Kuo Hsiang Chuang
Kaohsiung Medical University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Kuo Hsiang Chuang.
Bioconjugate Chemistry | 2012
Tian-Lu Cheng; Kuo Hsiang Chuang; Bing Mae Chen; Steve R. Roffler
Attachment of poly(ethylene glycol) (PEG) to proteins, peptides, liposomes, drugs, and nanoparticles can improve pharmaceutical pharmacokinetic properties and enhance in vivo biological efficacy. Since the first PEGylated product was approved by the Food and Drug Administration in 1990, increasing numbers of PEGylated compounds have entered clinical use. Successful clinical development of PEGylated pharmaceuticals requires accurate methods for the qualitative and quantitative analysis of intact PEG conjugates in biological fluids. In this article, we review assay methods that can be utilized for the detection and measurement of PEGylated pharmaceuticals in complex biological samples for determination of biodistribution and pharmacokinetic properties. In particular, we describe relevant colorimetric, chromatographic, radiolabeled, biological, and enzyme-linked immunosorbent assays for the pharmacokinetic study of PEGylated molecules.
Bioconjugate Chemistry | 2010
Yu Cheng Su; Bing Mae Chen; Kuo Hsiang Chuang; Tian-Lu Cheng; Steve R. Roffler
Poly(ethylene glycol) (PEG) is often attached to compounds to increase serum half-life, reduce immunogenicity, and enhance bioavailability. Accurate and sensitive quantification of PEG conjugates is critical for product development, pharmacokinetic measurements, and efficacy studies. However, PEGylated compounds can be difficult to quantify due to epitope masking by PEG. We previously generated two monoclonal antibodies to PEG (AGP3, IgM and E11, IgG) for quantitative detection of PEGylated proteins. We now report the identification of two second-generation mAbs to PEG (AGP4, IgM and 3.3, IgG) that bind to the repeating subunits of the PEG backbone and facilitate more sensitive quantification of a wider range of PEGylated compounds. A sandwich ELISA in which AGP4/3.3-biotin was employed as the capture/detection antibodies allowed quantification of PEG-Qdot 525 with 14-50-fold greater sensitivity than the original AGP3/E11 combination. Pegasys (PEG-interferon alpha-2a), PEG-Intron (PEG-interferon alpha-2b), Neulasta (PEG-G-CSF), and Lipo-Dox (PEGylated liposomal doxorubicin) could also be quantified with low ng/mL detection limits. The assay tolerated the presence of 50% human serum or 20% free PEG molecules. These new anti-PEG antibodies appear useful for qualitative and quantitative analysis of a wide range of PEGylated compounds.
Molecular Cancer Therapeutics | 2010
Kuo Hsiang Chuang; Hsin Ell Wang; Fang Ming Chen; Shey Cherng Tzou; Chiu Min Cheng; Ya Chen Chang; Wei-Lung Tseng; Jentaie Shiea; Shiu Ru Lin; Jaw-Yuan Wang; Bing Mae Chen; Steve R. Roffler; Tian-Lu Cheng
PEGylated nanoparticles and macromolecules are increasingly used in cancer imaging and anticancer treatment. The role of receptor-mediated endocytosis in the efficacy of these agents, however, has not been clearly defined. Here, we developed a matched pair of endocytic and nonendocytic receptors to directly and unambiguously assess this issue. The ligand-binding domains of the low-density lipoprotein receptor (LDLR) or a truncated LDLR lacking the NPXY endocytosis motif (ΔLDLR) were replaced with an anti–polyethylene glycol antibody (αPEG) to form endocytic αPEG-LDLR and nonendocytic αPEG-ΔLDLR receptors. The receptors were stably expressed at similar levels on the surface of HCC36 cells. HCC36/αPEG-LDLR cells, but not HCC36/αPEG-ΔLDLR cells, rapidly endocytosed PEG-quantum dots and PEG-liposomal doxorubicin (Lipo-Dox) in vitro and in vivo. Lipo-Dox was significantly more cytotoxic to HCC36/αPEG-LDLR cells than to HCC36/αPEG-ΔLDLR cells. HCC36/αPEG-LDLR tumors also accumulated significantly more PEGylated near-IR probes (PEG-NIR797) and PEG-liposomal-111In than HCC36/αPEG-ΔLDLR tumors in vivo. Furthermore, Lipo-Dox more significantly suppressed the growth of established HCC36/αPEG-LDLR tumors as compared with HCC36/αPEG-ΔLDLR tumors. Our data show that endocytosis of PEGylated probes and drugs enhances both cancer imaging and anticancer efficacy, indicating that endocytic receptors are superior targets for the design of cancer imaging probes and immunoliposomal drugs. Mol Cancer Ther; 9(6); 1903–12. ©2010 AACR.
Clinical Cancer Research | 2012
Chih Hung Chuang; Kuo Hsiang Chuang; Hsin Ell Wang; Steve R. Roffler; Jen Taie Shiea; Shey Cherng Tzou; Ta Chun Cheng; Chien Han Kao; Shih Yen Wu; Wei-Lung Tseng; Chiu Min Cheng; Ming Feng Hou; Ju Ming Wang; Tian-Lu Cheng
Purpose: To develop an imaging technology for protease activities in patients that could help in prognosis prediction and in design of personalized, protease-based inhibitors and prodrugs for targeted therapy. Experimental Design: Polyethylene glycol (PEG) was covalently attached to the N-terminus of a hydrophilic peptide substrate (GPLGVR) for matrix metalloproteinase (MMP) to increase hydrophilicity. PEG-peptide was then linked to a hydrophobic tetramethylrhodamine (TMR) domain and labeled with 18F to form a PEG-peptide-18F-TMR probe. Specific cleavage of the probe by MMP2 was tested in vitro by matrix-assisted laser desorption/ionization–time-of-flight (MALDI-TOF). In vivo imaging of MMP2-expressing tumors was evaluated by micro-PET. Results: The hydrophobic TMR fragment (948 Da) was specifically generated by MMP2 enzymes and MMP-expressing HT1080 cells but not control MCF-7 cells. MMP-expressing HT1080 cells and tumors selectively accumulated the hydrolyzed, hydrophobic TMR fragment at sites of protease activity. Importantly, we found that 18F-labeled probe (18F-TMR) preferentially localized in HT1080 tumors but not control MCF-7 tumors as shown by micro-PET. Uptake of the probe in HT1080 tumors was 18.4 ± 1.9-fold greater than in the MCF-7 tumors 30 minutes after injection. These results suggest that the PEG-peptide-18F-TMR probe displays high selectivity for imaging MMP activity. Conclusions: This strategy successfully images MMP expression in vivo and may be extended to other proteases to predict patient prognosis and to design personalized, protease-based inhibitors and prodrug-targeted therapies. Clin Cancer Res; 18(1); 238–47. ©2011 AACR.
The Journal of Nuclear Medicine | 2010
Kuo Hsiang Chuang; Hsin Ell Wang; Ta Chun Cheng; Shey Cherng Tzou; Wei-Lung Tseng; Wen Chun Hung; Ming Hong Tai; Tien Kuei Chang; Steve R. Roffler; Tian-Lu Cheng
A reporter gene can provide important information regarding the specificity and efficacy of gene or cell therapies. Although reporter genes are increasingly used in experimental and clinical studies, a highly specific yet nonimmunogenic reporter that can track genes and cells in vivo by multiple imaging technologies still awaits development. In this study, we constructed a versatile and nonimmunogenic reporter gene to noninvasively image gene expression or cell delivery by optical imaging, MRI, and small-animal PET. Methods: We cloned and expressed a membrane-anchored anti–polyethylene glycol (PEG) reporter that consists of the Fab fragment of a mouse anti-PEG monoclonal antibody, AGP3, fused to the C-like extracellular-transmembrane-cytosolic domains of the mouse B7-1 receptor. Binding of PEGylated probes (PEG-NIR797 for optical imaging, PEG–superparamagnetic iron oxide for MRI, and 124I-PEG for small-animal PET) were examined in vitro and in vivo. In addition, we compared the specificity, immunogenicity, and probe toxicity of the anti-PEG reporter with the gold standard reporter gene, type 1 herpes simplex virus thymidine kinase (HSV-tk). Finally, we derived a humanized anti-PEG reporter and evaluated its imaging function in vivo with subcutaneous and metastatic tumor models in mice. Results: The cells or tumors that stably expressed anti-PEG reporters selectively accumulated various PEGylated imaging probes and could be detected by optical imaging, MRI, and small-animal PET. Importantly, the anti-PEG reporter displayed an imaging specificity comparable to the HSV-tk reporter but did not provoke immune responses or cause toxicity to the host. Furthermore, the humanized anti-PEG reporter retained high imaging specificity in vivo. Conclusion: The highly specific and nonimmunogenic anti-PEG reporter may be paired with PEGylated probes to provide a valuable system to image gene expression or cell delivery in experimental and clinical studies.
Biomaterials | 2014
Chien Han Kao; Jaw-Yuan Wang; Kuo Hsiang Chuang; Chih Hung Chuang; Ta Chun Cheng; Yuan Chin Hsieh; Yun long Tseng; Bing Mae Chen; Steve R. Roffler; Tian-Lu Cheng
Methoxy PEGylated nanoparticles (mPEG-NPs) are increasingly used for cancer imaging and therapy. Here we describe a general and simple approach to confer tumor tropism to any mPEG-NP. We demonstrate this approach with humanized bispecific antibodies (BsAbs) that can bind to both mPEG molecules on mPEG-NPs and to EGFR or HER2 molecules overexpressed on the surface of cancer cells. Simple mixing of BsAbs with mPEG-NPs can mediate preferential binding of diverse mPEG-NPs to cancer cells that overexpress EGFR or HER2 under physiological conditions and significantly increase cancer cell killing by liposomal doxorubicin to EGFR(+) and HER2(+) cancer cells. BsAbs modification also enhanced accumulation of fluorescence-labeled NPs and significantly increased the anticancer activity of drug-loaded NPs to antigen-positive human tumors in a mouse model. Anti-mPEG BsAbs offer a simple one-step method to confer tumor specificity to mPEG-NPs for enhanced tumor accumulation and improved therapeutic efficacy.
Molecular Cancer Therapeutics | 2015
Hsin Yi Tung; Yu Cheng Su; Bing Mae Chen; Pierre Alain Burnouf; Wei-Chiao Huang; Kuo Hsiang Chuang; Yu-Ting Yan; Tian-Lu Cheng; Steve R. Roffler
Polyethylene glycol (PEG) is attached to many peptides, proteins, liposomes, and nanoparticles to reduce their immunogenicity and improve their pharmacokinetic and therapeutic properties. Here, we describe hybrid antibodies that can selectively deliver PEGylated medicines, imaging agents, or nanomedicines to target cells. Human IgG1 hybrid antibodies αPEG:αHER2 and αPEG:αCD19 were shown by ELISA, FACS, and plasmon resonance to bind to both PEG and HER2 receptors on SK-BR-3 breast adenocarcinoma and BT-474 breast ductal carcinoma cells or CD19 receptors on Ramos and Raji Burkitts lymphoma cells. In addition, αPEG:αHER2 specifically targeted PEGylated proteins, liposomes, and nanoparticles to SK-BR-3 cells that overexpressed HER2, but not to HER2-negative MCF-7 breast adenocarcinoma cells. Endocytosis of PEGylated nanoparticles into SK-BR-3 cells was induced specifically by the αPEG:αHER2 hybrid antibody, as observed by confocal imaging of the accumulation of Qdots inside SK-BR-3 cells. Treatment of HER2+ SK-BR-3 and BT-474 cancer cells with αPEG:αHER2 and the clinically used chemotherapeutic agent PEGylated liposomal doxorubicin for 3 hours enhanced the in vitro effectiveness of PEGylated liposomal doxorubicin by over two orders of magnitude. Hybrid anti-PEG antibodies offer a versatile and simple method to deliver PEGylated compounds to cellular locations and can potentially enhance the therapeutic efficacy of PEGylated medicines. Mol Cancer Ther; 14(6); 1317–26. ©2015 AACR.
The Scientific World Journal | 2015
Ta Chun Cheng; Kuo Hsiang Chuang; Steve R. Roffler; Kai Wen Cheng; Yu Lin Leu; Chih Hung Chuang; Chien Chaio Huang; Chien Han Kao; Yuan Chin Hsieh; Long Sen Chang; Tian-Lu Cheng; Chien Shu Chen
Glucuronidation is a major metabolism process of detoxification for carcinogens, 4-(methylnitrosamino)-1-(3-pyridy)-1-butanone (NNK) and 1,2-dimethylhydrazine (DMH), of reactive oxygen species (ROS). However, intestinal E. coli β-glucuronidase (eβG) has been considered pivotal to colorectal carcinogenesis. Specific inhibition of eβG may prevent reactivating the glucuronide-carcinogen and protect the intestine from ROS-mediated carcinogenesis. In order to develop specific eβG inhibitors, we found that 59 candidate compounds obtained from the initial virtual screening had high inhibition specificity against eβG but not human βG. In particular, we found that compounds 7145 and 4041 with naphthalenylidene-benzenesulfonamide (NYBS) are highly effective and selective to inhibit eβG activity. Compound 4041 (IC50 = 2.8 μM) shows a higher inhibiting ability than compound 7145 (IC50 = 31.6 μM) against eβG. Furthermore, the molecular docking analysis indicates that compound 4041 has two hydrophobic contacts to residues L361 and I363 in the bacterial loop, but 7145 has one contact to L361. Only compound 4041 can bind to key residue (E413) at active site of eβG via hydrogen-bonding interactions. These novel NYBS-based eβG specific inhibitors may provide as novel candidate compounds, which specifically inhibit eβG to reduce eβG-based carcinogenesis and intestinal injury.
Nature Communications | 2017
Yu Cheng Su; Pierre Alain Burnouf; Kuo Hsiang Chuang; Bing Mae Chen; Tian-Lu Cheng; Steve R. Roffler
Triple-negative breast cancer (TNBC) lacks effective treatment options due to the absence of traditional therapeutic targets. The epidermal growth factor receptor (EGFR) has emerged as a promising target for TNBC therapy because it is overexpressed in about 50% of TNBC patients. Here we describe a PEG engager that simultaneously binds polyethylene glycol and EGFR to deliver PEGylated nanomedicines to EGFR+ TNBC. The PEG engager displays conditional internalization by remaining on the surface of TNBC cells until contact with PEGylated nanocarriers triggers rapid engulfment of nanocargos. PEG engager enhances the anti-proliferative activity of PEG-liposomal doxorubicin to EGFR+ TNBC cells by up to 100-fold with potency dependent on EGFR expression levels. The PEG engager significantly increases retention of fluorescent PEG probes and enhances the antitumour activity of PEGylated liposomal doxorubicin in human TNBC xenografts. PEG engagers with specificity for EGFR are promising for improved treatment of EGFR+ TNBC patients.
mAbs | 2014
Yu Cheng Su; Talal S. Al-Qaisi; Hsin Yi Tung; Tian-Lu Cheng; Kuo Hsiang Chuang; Bing Mae Chen; Steve R. Roffler
Modification of antibody class and binding properties typically requires cloning of antibody genes, antibody library construction, phage or yeast display and recombinant antibody expression. Here, we describe an alternative “cloning-free” approach to generate antibodies with altered antigen-binding and heavy chain isotype by mimicking the germinal center reaction in antibody-secreting hybridoma cells. This was accomplished by lentiviral transduction and controllable expression of activation-induced cytidine deaminase (AID) to generate somatic hypermutation and class switch recombination in antibody genes coupled with high-throughput fluorescence-activated cell sorting (FACS) of hybridoma cells to detect altered antibody binding properties. Starting from a single established hybridoma clone, we isolated mutated antibodies that bind to a low-temperature structure of polyethylene glycol (PEG), a polymer widely used in nanotechnology, biotechnology and pharmaceuticals. FACS of AID-infected hybridoma cells also facilitated rapid identification of class switched variants of monoclonal IgM to monoclonal IgG. Mimicking the germinal center reaction in hybridoma cells may offer a general method to identify and isolate antibodies with altered binding properties and class-switched heavy chains without the need to carry out DNA library construction, antibody engineering and recombinant protein expression.